Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros











Intervalo de año de publicación
1.
Xenobiotica ; 51(2): 222-238, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33078965

RESUMEN

Dilated cardiomyopathy (DCM) is a disease of the myocardium defined by left ventricular enlargement and systolic dysfunction leading to heart failure. Danicamtiv, a new targeted myosin activator designed for the treatment of DCM, was characterised in in vitro and in vivo preclinical studies. Danicamtiv human hepatic clearance was predicted to be 0.5 mL/min/kg from in vitro metabolic stability studies in human hepatocytes. For human, plasma protein binding was moderate with a fraction unbound of 0.16, whole blood-to-plasma partitioning ratio was 0.8, and danicamtiv showed high permeability and no efflux in a Caco-2 cell line. Danicamtiv metabolism pathways in vitro included CYP-mediated amide-cleavage, N-demethylation, as well as isoxazole- and piperidine-ring-opening. Danicamtiv clearance in vivo was low across species with 15.5, 15.3, 1.6, and 5.7 mL/min/kg in mouse, rat, dog, and monkey, respectively. Volume of distribution ranged from 0.24 L/kg in mouse to 1.7 L/kg in rat. Oral bioavailability ranged from 26% in mouse to 108% in dog. Simple allometric scaling prediction of human plasma clearance, volume of distribution, and half-life was 0.64 mL/min/kg, 0.98 L/kg, and 17.7 h, respectively. Danicamtiv preclinical attributes and predicted human pharmacokinetics supported advancement toward clinical development.


Asunto(s)
Cardiomiopatía Dilatada/tratamiento farmacológico , Animales , Disponibilidad Biológica , Células CACO-2 , Perros , Hepatocitos , Humanos , Masculino , Ratones , Microsomas Hepáticos , Miosinas , Unión Proteica , Ratas
2.
Expert Opin Drug Metab Toxicol ; 11(8): 1281-302, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26005795

RESUMEN

INTRODUCTION: A number of withdrawn drugs are known to undergo bioactivation by a range of drug metabolizing enzymes to chemically reactive metabolites that bind covalently to protein and DNA resulting in organ toxicity and carcinogenesis, respectively. An important goal in drug discovery is to identify structural sites of bioactivation within discovery molecules for providing strategic modifications that eliminate or minimize reactive metabolite formation, while maintaining target potency, selectivity and desired pharmacokinetic properties leading to the development of efficacious and nontoxic drugs. AREAS COVERED: This review covers experimental techniques currently used to detect reactive drug metabolites and provides recent examples where information from mechanistic in vitro studies was successfully used to redesign candidate drugs leading to blocked or minimized bioactivation. Reviewed techniques include in vitro radiolabeled drug covalent binding to protein and reactive metabolite trapping with reagents such as glutathione, cyanide, semicarbazide and DNA bases. Case studies regarding reactive metabolite detection using a combination of varied techniques, including liquid chromatography-tandem mass spectrometry and NMR analyses and subsequent structural modification are discussed. EXPERT OPINION: Information derived from state-of-art mechanistic drug metabolism studies can be used successfully to direct medicinal chemistry towards the synthesis of candidate drugs devoid of bioactivation liabilities, while maintaining desired pharmacology and pharmacokinetic properties.


Asunto(s)
Diseño de Fármacos , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/metabolismo , Preparaciones Farmacéuticas/metabolismo , Animales , Cromatografía Liquida/métodos , ADN/metabolismo , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/prevención & control , Humanos , Espectroscopía de Resonancia Magnética/métodos , Unión Proteica , Espectrometría de Masas en Tándem
3.
Drug Metab Dispos ; 41(1): 111-21, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23052971

RESUMEN

Ibuprofen is metabolized to chemically reactive acyl glucuronide and S-acyl-CoA metabolites that are proposed to transacylate glutathione (GSH) forming ibuprofen-S-acyl-GSH (I-SG) in vivo. Herein, we report the detection of novel metabolites of ibuprofen, namely ibuprofen-N-acyl-cysteinylglycine (I-N-CG), ibuprofen-N-acyl-cysteine (I-N-C), and the mercapturic acid conjugate, ibuprofen-S-acyl-N-acetylcysteine (I-S-NAC), in urine from an ibuprofen-dosed volunteer. Thus, analysis of ibuprofen-dosed (Advil, 800 mg, Pfizer, Madison, NJ) human urine extracts by sensitive liquid chromatography tandem mass spectrometric detection resulted in the identification of I-N-CG, I-N-C, and I-S-NAC derivatives as minor metabolites (6.0, 1.7, and 0.2 µg excreted 10-hours postadministration, respectively). I-N-CG is proposed to be formed from the degradation of I-SG by γ-glutamyltranspeptidase (γ-GT)-mediated cleavage of the γ-glutamyl group, leading to an unstable ibuprofen-S-acyl-cysteinylglycine (I-S-CG) intermediate that undergoes spontaneous S to N intramolecular rearrangement. Then, dipeptidase-mediated cleavage of glycine from I-N-CG leads to the formation of I-N-C. Treatment of racemic I-SG (100 µM) in vitro with commercially available bovine kidney γ-GT (0.1 units/ml) in buffer at pH 7.4 and 37°C resulted in its complete degradation, yielding (R)- and (S)-I-N-CG after 15 minutes of incubation. In vitro enzyme kinetic studies with bovine kidney γ-GT incubated separately with (R)- and (S)-I-SG isomers revealed no enantioselective degradation. Results from these studies provided evidence that ibuprofen is metabolized in human to reactive transacylating-type intermediates that react with GSH, forming I-SG thioester that, following degradation by γ-GT and dipeptidase enzymes and following S to N intramolecular rearrangement, leads to the urinary excretion of the I-N-CG and I-N-C amide-linked conjugates, respectively.


Asunto(s)
Glutatión/análogos & derivados , Ibuprofeno/análogos & derivados , gamma-Glutamiltransferasa/metabolismo , Animales , Bovinos , Cromatografía Liquida , Glutatión/metabolismo , Humanos , Ibuprofeno/metabolismo , Técnicas In Vitro , Espectroscopía de Resonancia Magnética , Espectrometría de Masas en Tándem
4.
Drug Metab Dispos ; 40(8): 1515-26, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22577085

RESUMEN

Carboxylic acid-containing nonsteroidal anti-inflammatory drugs (NSAIDs) can be metabolized to chemically reactive acyl glucuronide and/or S-acyl-CoA thioester metabolites capable of transacylating GSH. We investigated the metabolism of the NSAID mefenamic acid (MFA) to metabolites that transacylate GSH, leading to MFA-S-acyl-GSH thioester (MFA-SG) formation in incubations with rat and human hepatocytes and in vivo in rat bile. Thus, incubation of MFA (1-500 µM) with rat hepatocytes led to the detection of MFA-1-ß-O-acyl glucuronide (MFA-1-ß-O-G), MFA-S-acyl-CoA (MFA-SCoA), and MFA-SG by liquid chromatography-tandem mass spectrometric analysis. The C(max) of MFA-SG (330 nM; 10-min incubation with 100 µM MFA) was 120- to 1400-fold higher than the C(max) of drug S-acyl-GSH adducts detected from studies with other carboxylic acid drugs to date. MFA-SG was also detected in incubations with human hepatocytes, but at much lower concentrations. Inhibition of MFA acyl glucuronidation in rat hepatocytes had no effect on MFA-SG formation, whereas a 58 ± 1.7% inhibition of MFA-SCoA formation led to a corresponding 66 ± 3.5% inhibition of MFA-SG production. Reactivity comparisons with GSH in buffer showed MFA-SCoA to be 80-fold more reactive than MFA-1-ß-O-G forming MFA-SG. MFA-SG was detected in MFA-dosed (100 mg/kg) rat bile, where 17.4 µg was excreted after administration. In summary, MFA exhibited bioactivation in rat and human hepatocytes and in vivo in rat, leading to reactive acylating derivatives that transacylate GSH. The formation of MFA-SG in hepatocytes was shown not to be mediated by reaction with MFA-1-ß-O-G, and not solely by MFA-SCoA, but perhaps also by intermediary MFA-acyl-adenylate formation, which is currently under investigation.


Asunto(s)
Biotransformación , Glutatión/farmacocinética , Ácido Mefenámico/farmacocinética , Animales , Cromatografía Liquida , Hepatocitos/metabolismo , Humanos , Técnicas In Vitro , Ratas , Espectrometría de Masas en Tándem
5.
Drug Metab Dispos ; 39(12): 2387-94, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21865320

RESUMEN

Sandwich-cultured rat hepatocytes are used in drug discovery for pharmacological and toxicological assessment of drug candidates, yet their utility as a functional model for drug transporters has not been fully characterized. To evaluate the system as an in vitro model for drug transport, expression changes of hepatic transporters relative to whole liver and freshly isolated hepatocytes (day 0) were examined by real-time quantitative reverse transcription-polymerase chain reaction for 4 consecutive days of culture. No significant differences in transporter expression levels were observed between freshly isolated hepatocytes and whole liver. Two distinct mRNA profiles were detected over time showing 1) a more than 5-fold decline in levels of uptake transporters such as Na(+)-taurocholate cotransporting polypeptide (Ntcp), organic anion transporter (Oat) 2, organic anion-transporting polypeptide (Oatp) 1a1, Oatp1a4, and Oatp1b2 and 2) a greater than 5-fold increase of efflux transporters P-glycoprotein (P-gp), breast cancer resistance protein (Bcrp), and multidrug resistance-related proteins (Mrp) 1, 2, 3, and 4. In addition, protein levels and functional activities for selected transporters were also determined. Protein levels for Mrp2, Bcrp, P-gp, Ntcp, and Oatp1a4 corresponded to changes in mRNA. Functional activities of Oatps and Oct1 exhibited a 3- and 4-fold decrease on day 2 and day 4, respectively, relative to that on day 0, whereas a more than 10-fold reduction in Oat2 activity was observed. These results indicate that the cell culture conditions used herein did not provide an optimal environment for expression of all hepatic transporters. Significant time-dependent alterations in basal gene expression patterns of transporters were detected compared with those in liver or freshly isolated hepatocytes. Further work and new strategies are required to improve the validity of this model as an in vitro tool for in vivo drug transport or biliary clearance prediction.


Asunto(s)
Proteínas Portadoras/metabolismo , Hepatocitos/metabolismo , Preparaciones Farmacéuticas/metabolismo , Animales , Células Cultivadas , Masculino , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
6.
Chem Res Toxicol ; 24(7): 992-1002, 2011 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-21506562

RESUMEN

Aqueous kava root preparations have been consumed in the South Pacific as an apparently safe ceremonial and cultural drink for centuries. However, several reports of hepatotoxicity have been linked to the consumption of kava extracts in Western countries, where mainly ethanolic or acetonic extracts are used. The mechanism of toxicity has not been established, although several theories have been put forward. The composition of the major constituents, the kava lactones, varies according to preparation method and species of kava plant, and thus, the toxicity of the individual lactones has been tested in order to establish whether a single lactone or a certain composition of lactones may be responsible for the increased prevalence of kava-induced hepatotoxicity in Western countries. However, no such conclusion has been made on the basis of current data. Inhibition or induction of the major metabolizing enzymes, which might result in drug interactions, has also gained attention, but ambiguous results have been reported. On the basis of the chemical structures of kava constituents, the formation of reactive metabolites has also been suggested as an explanation of toxicity. Furthermore, skin rash is a side effect in kava consumers, which may be indicative of the formation of reactive metabolites and covalent binding to skin proteins leading to immune-mediated responses. Reactive metabolites of kava lactones have been identified in vitro as glutathione (GSH) conjugates and in vivo as mercapturates excreted in urine. Addition of GSH to kava extracts has been shown to reduce cytotoxicity in vitro, which suggests the presence of inherently reactive constituents. Only a few studies have investigated the toxicity of the minor constituents present in kava extract, such as pipermethystine and the flavokavains, where some have been shown to display higher in vitro cytotoxicity than the lactones. To date, there remains no indisputable reason for the increased prevalence of kava-induced hepatotoxicity in Western countries.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Kava/química , Animales , Sistema Enzimático del Citocromo P-450/genética , Sistema Enzimático del Citocromo P-450/metabolismo , Exantema/inducido químicamente , Exantema/patología , Glutatión/metabolismo , Humanos , Lactonas/efectos adversos , Lactonas/química , Lactonas/toxicidad , Raíces de Plantas/química
7.
Curr Drug Metab ; 12(3): 229-44, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20946099

RESUMEN

Carboxylic acid-containing drugs can be metabolized to chemically-reactive acyl glucuronide, S-acyl-CoA thioester, and/or intermediate acyl-adenylate metabolites that are capable of transacylating the cysteinyl-thiol of glutathione (GSH) resulting in the formation of drug-S-acyl-GSH thioesters detected in-vivo in bile and in-vitro in hepatocytes. Authentic S-acyl-GSH thioesters of carboxylic acids can be readily synthesized by modifying the cysteinyl-thiol group of GSH with an applicable acylating reagent. Bionanalytical characterization of S-acyl-GSH derivatives has demonstrated enhanced extraction efficiency from biological samples when formic acid is included in appropriate extraction solvents, and that tandem mass spectrometry of S-acyl-GSH conjugates results in fragmentation producing a common MH+-147 Da product ion. Chemical reactivity comparisons have shown that S-acyl-CoA thioester and acyl-adenylate conjugates are more reactive than their corresponding 1-ß-O-acyl glucuronides toward the transacylation of GSH forming S-acyl-GSH thioesters. S-Acyl-GSH thioester derivatives are also chemically-reactive electrophiles capable of transacylating biological nucleophiles. Glutathione S-transferases (GSTs) weakly catalyze S-acyl-GSH conjugate formation from S-acyl-CoA, acyl-adenylate, and 1-ß-O-acyl glucuronide substrates; however purified-GSTs have also been shown to hydrolyze S-acyl-GSH thioesters. Mechanistic in vitro studies in hepatocytes have revealed the primary importance of the S-acyl-CoA formation pathway leading to S-acyl-GSH-adduct formation. In addition to being hydrolytically-unstable in hepatocytes and plasma, S-acyl-GSH thioesters undergo γ-glutamyltranspeptidase-mediated cleavage of the γ-glutamyl-group leading to N-acyl-cysteinylglycine amide-linked products. In summary, S-acyl GSH thioesters are indicators of reactive transacylating metabolite formation produced from the biotransformation of carboxylic acids, but since they are also chemically-reactive, perhaps these derivatives can contribute to covalent binding to tissue proteins and potential toxicity.


Asunto(s)
Glutatión/análogos & derivados , Acilcoenzima A/química , Acilcoenzima A/metabolismo , Animales , Glutatión/química , Glutatión/metabolismo , Glutatión/farmacología , Humanos , Unión Proteica , Proteínas/metabolismo , Sulfuros/química , Sulfuros/metabolismo
8.
Drug Metab Dispos ; 38(1): 133-42, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19786506

RESUMEN

Flunoxaprofen (FLX) is a chiral nonsteroidal anti-inflammatory drug that was withdrawn from clinical use because of concerns of potential hepatotoxicity. FLX undergoes highly stereoselective chiral inversion mediated through the FLX-S-acyl-CoA thioester (FLX-CoA) in favor of the (R)-(-)-isomer. Acyl-CoA thioester derivatives of acidic drugs are chemically reactive species that are known to transacylate protein nucleophiles and glutathione (GSH). In this study, we investigated the relationship between the stereoselective metabolism of (R)-(-)- and (S)-(+)-FLX to FLX-CoA and the subsequent transacylation of GSH forming FLX-S-acyl-glutathione (FLX-SG) in incubations with rat hepatocytes in suspension. Thus, when hepatocytes (2 million cells/ml) were treated with (R)-(-)- or (S)-(+)-FLX (100 microM), both FLX-CoA and FLX-SG were detected by sensitive liquid chromatography-tandem mass spectrometry techniques. However, these derivatives were observed primarily from (R)-(-)-FLX incubation extracts, for which the formation rates of FLX-CoA and FLX-SG were rapid, reaching maximum concentrations of 42 and 2.8 nM, respectively, after 6 min of incubation. Incubations with (S)-(+)-FLX over 60 min displayed 8.1 and 2.7% as much FLX-CoA and FLX-SG area under the concentration versus time curves, respectively, compared with corresponding incubations with (R)-(-)-FLX. Coincubation of lauric acid (1000 microM) with (R)-(-)-FLX (10 microM) led to the complete inhibition of FLX-CoA formation and a 98% inhibition of FLX-SG formation. Reaction of authentic (R,S)-FLX-CoA (2 microM) with GSH (10 mM) in buffer (pH 7.4, 37 degrees C) showed the quantitative formation of FLX-SG after 3 h of incubation. Together, these results demonstrate the stereoselective transacylation of GSH in hepatocyte incubations containing (R)-(-)-FLX, which is consistent with bioactivation by stereoselective (R)-FLX-CoA formation.


Asunto(s)
Acilcoenzima A/biosíntesis , Benzoxazoles/química , Benzoxazoles/metabolismo , Ésteres/metabolismo , Glutatión/metabolismo , Hepatocitos/metabolismo , Compuestos de Sulfhidrilo/metabolismo , Acilcoenzima A/metabolismo , Acilación , Animales , Antiinflamatorios no Esteroideos/metabolismo , Antiinflamatorios no Esteroideos/farmacocinética , Benzoxazoles/farmacocinética , Biocatálisis , Biotransformación , Canfanos/farmacología , Cromatografía Líquida de Alta Presión , Inhibidores Enzimáticos/farmacología , Ácido Glucurónico/metabolismo , Glutatión/química , Hepatocitos/efectos de los fármacos , Hepatocitos/enzimología , Ibuprofeno/metabolismo , Cinética , Ácidos Láuricos/farmacología , Masculino , Estructura Molecular , Ratas , Ratas Sprague-Dawley , Estereoisomerismo , Espectrometría de Masas en Tándem
9.
Drug Metab Dispos ; 37(5): 1073-82, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19196839

RESUMEN

Phenylacetic acid (PAA) represents a substructure of a class of nonsteroidal anti-inflammatory carboxylic acid-containing drugs capable of undergoing metabolic activation in the liver to acylcoenzyme A (CoA)- and/or acyl glucuronide-linked metabolites that are proposed to be associated with the formation of immunogenic, and hence potentially hepatotoxic, drug-protein adducts. Herein, we investigated the ability of PAA to undergo phenylacetyl-S-acyl-CoA thioester (PA-CoA)-mediated covalent binding to protein in incubations with freshly isolated rat hepatocytes in suspension. Thus, when hepatocytes were incubated with phenylacetic acid carboxy-(14)C (100 microM) and analyzed for PA-CoA formation and covalent binding of PAA to protein and over a 3-h time period, both PA-CoA formation and covalent binding to protein increased rapidly, reaching 1.3 microM and 291 pmol equivalents/mg protein after 4 and 6 min of incubation, respectively. However, the covalent binding of PAA to protein was reversible and decreased by 72% at the 3-h time point. After 3 h of incubation, PAA was shown to be metabolized primarily to phenylacetyl-glycine amide (84%). No PAA-acyl glucuronide was detected in the incubation extracts. PA-CoA reacted readily with glutathione in buffer, forming PA-S-acyl-glutathione; however, this glutathione conjugate was not detected in hepatocyte incubation extracts. Coincubation of hepatocytes with lauric acid led to a marked inhibition of PA-CoA formation and a corresponding inhibition of covalent binding to protein. SDS-polyacrylamide gel electrophoresis analysis showed the formation of two protein adducts having molecular masses of approximately 29 and approximately 33 kDa. In summary, PA-CoA formation in rat hepatocytes leads to the highly selective, but reversible, covalent binding to hepatocyte proteins, but not to the transacylation of glutathione.


Asunto(s)
Hepatocitos/metabolismo , Fenilacetatos/metabolismo , Animales , Biotransformación , Separación Celular , Cromatografía Liquida , Electroforesis en Gel de Poliacrilamida , Glutatión/metabolismo , Técnicas In Vitro , Ácidos Láuricos/farmacología , Espectroscopía de Resonancia Magnética , Microsomas Hepáticos/metabolismo , Peso Molecular , Unión Proteica , Ratas , Ratas Sprague-Dawley , Espectrometría de Masas en Tándem
10.
Chem Res Toxicol ; 21(9): 1749-59, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18680316

RESUMEN

Ibuprofen is metabolized to chemically reactive ibuprofen-1- O-acyl-glucuronide (I-1- O-G) and ibuprofen- S-acyl-CoA (I-CoA) derivatives, which are proposed to mediate the formation of drug-protein adducts via the transacylation of protein nucleophiles. We examined the ability of ibuprofen to undergo enantioselective metabolism to ibuprofen- S-acyl-glutathione thioester (I-SG) in incubations with rat hepatocytes, where I-CoA formation is known to be highly enantioselective in favor of the (R)-(-)-ibuprofen isomer. We proposed that potential enantioselective transacylation of glutathione forming I-SG in favor of the (R)-(-)-isomer would reveal the importance of acyl-CoA formation, versus acyl glucuronidation, in the generation of reactive transacylating-type intermediates of the drug. Thus, when (R)-(-)- and (S)-(+)-ibuprofen (100 microM) were incubated with hepatocytes, the presence of I-CoA and I-SG was detected in incubation extracts by LC-MS/MS techniques. The formation of I-CoA and I-SG in hepatocyte incubations with (R)-(-)-ibuprofen was rapid and reached maximum concentrations of 2.6 microM and 1.3 nM, respectively, after 8-10 min of incubation. By contrast, incubations with (S)-(+)-ibuprofen resulted in 8% and 3.9% as much I-CoA and I-SG formation, respectively, compared to that in corresponding incubations with the (R)-(-)-isomer. Experiments with a pseudoracemic mixture of (R)-(-)-[3,3,3-(2)H3]- and (S)-(+)-ibuprofen showed that >99% of the I-SG detected in hepatocyte incubations contained deuterium and therefore was derived primarily from (R)-(-)-ibuprofen bioactivation. Inhibition of (R)-(-)-ibuprofen (10 microM) glucuronidation with (-)-borneol (100 microM) led to a 98% decrease in I-1-O-G formation; however, no decrease in I-SG production was observed. Coincubation with pivalic, valproic, or lauric acid (500 microM each) was shown to lead to a significant inhibition of I-CoA formation and a corresponding decrease in I-SG production. Results from these studies demonstrate that the reactive I-CoA derivative, and not the I-1-O-G metabolite, plays a central role in the transacylation of GSH in incubations with rat hepatocytes.


Asunto(s)
Glutatión/análogos & derivados , Hepatocitos/metabolismo , Ibuprofeno/análogos & derivados , Ibuprofeno/metabolismo , Animales , Canfanos/farmacología , Cromatografía Liquida , Glutatión/antagonistas & inhibidores , Glutatión/biosíntesis , Glutatión/química , Hepatocitos/química , Ibuprofeno/antagonistas & inhibidores , Ibuprofeno/química , Ácidos Láuricos/farmacología , Masculino , Espectrometría de Masas , Conformación Molecular , Ácidos Pentanoicos/farmacología , Ratas , Ratas Sprague-Dawley , Estereoisomerismo , Ésteres del Ácido Sulfúrico/antagonistas & inhibidores , Ésteres del Ácido Sulfúrico/química , Ésteres del Ácido Sulfúrico/metabolismo , Factores de Tiempo , Ácido Valproico/farmacología
11.
Chem Res Toxicol ; 21(10): 1933-8, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18693772

RESUMEN

Diclofenac, a nonsteroidal antiinflammatory drug, is known to be metabolized to chemically reactive intermediates that transacylate GSH forming diclofenac-S-acyl-glutathione (D-SG) in vivo in rat and in vitro in rat and human hepatocytes. Recently, it was reported that the treatment of rats with diclofenac led to a substantial decrease in the activity of hepatic gamma-glutamyltranspeptidase (gamma-GT), an extracellular canalicular membrane enzyme. Because studies have indicated that D-SG is a chemically reactive transacylating species that is excreted into rat bile, we propose that D-SG formed in the liver may be a substrate for, and potential inhibitor of, hepatic gamma-GT. The present experiments were performed to investigate the ability of D-SG to be a substrate for gamma-GT in vivo in rat and in vitro with commercially available gamma-GT enzyme. We also examined the ability of D-SG to inhibit gamma-GT in vitro. Thus, LC-MS/MS analysis of bile extracts from diclofenac-dosed rats (200 mg/kg, iv) showed the presence of the gamma-GT-mediated D-SG degradation product diclofenac-N-acyl-cysteinylglycine (D- N-CG), where a total of approximately 8 microg was excreted 6 h postadministration. When D-SG (100 microM) was incubated with gamma-GT (1 unit/mL), the GSH adduct was degraded in a linear time-dependent fashion where approximately 94 microM D- N-CG was formed after 20 min of incubation. Dialysis studies showed that inhibition of gamma-GT by D-SG was completely reversible. Further inhibition studies showed that D-SG is a competitive inhibitor of the gamma-GT enzyme. Results from theses studies indicate that D-SG is a substrate for gamma-GT; however, the conjugate may not contribute significantly to the decrease in gamma-GT activity reported to occur in vivo in rat.


Asunto(s)
Diclofenaco/análogos & derivados , Glutatión/análogos & derivados , gamma-Glutamiltransferasa/metabolismo , Animales , Bilis/metabolismo , Diclofenaco/química , Diclofenaco/metabolismo , Dipéptidos/metabolismo , Glutatión/química , Glutatión/metabolismo , Estructura Molecular , Ratas , Ratas Sprague-Dawley , Espectrometría de Masas en Tándem
12.
Chem Res Toxicol ; 16(11): 1410-7, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14615966

RESUMEN

Diclofenac, a nonsteroidal anti-inflammatory drug, is metabolized to diclofenac-1-O-acyl glucuronide (D-1-O-G), a chemically reactive conjugate that has been implicated as playing a role in the idiosyncratic hepatoxicity associated with its use. The present studies investigated the ability of diclofenac to be metabolized to diclofenac-S-acyl-glutathione thioester (D-SG) in vitro in incubations with rat and human hepatocytes and whether its formation is dependent on a transacylation-type reaction between D-1-O-G and glutathione. When diclofenac (100 microM) was incubated with hepatocytes, D-SG was detected in both rat and human incubation extracts by a sensitive LC-MS/MS technique. The initial formation rate of D-SG in rat and human hepatocyte incubations was rapid and reached maximum concentrations of 1 and 0.8 nM, respectively, after 4 min of incubation. By contrast, during incubations with rat hepatocytes, the formation of D-1-O-G increased over 30 min of incubation, reaching a maximum concentration of 14.6 microM. Co-incubation of diclofenac (50 microM) with (-)-borneol (400 microM), an inhibitor of glucuronidation, led to a 94% decrease in D-1-O-G formation, although no significant decrease in D-SG production was observed. Together, these results indicate that diclofenac becomes metabolically activated in vitro in rat and human hepatocytes to reactive acylating derivatives that transacylate glutathione forming D-SG, but which is not solely dependent on transacylation by the D-1-O-G metabolite. From these results, it is proposed that reactive acylating metabolites of diclofenac, besides D-1-O-G, may be significant in the protein acylation that occurs in vivo and therefore also be important with regard to the mechanism(s) of diclofenac-mediated idiosyncratic hepatotoxicity.


Asunto(s)
Diclofenaco/análogos & derivados , Diclofenaco/farmacocinética , Glutatión/análogos & derivados , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Acilación , Animales , Antiinflamatorios no Esteroideos/metabolismo , Biotransformación , Canfanos/metabolismo , Canfanos/farmacología , Células Cultivadas , Cromatografía Líquida de Alta Presión/métodos , Diclofenaco/metabolismo , Relación Dosis-Respuesta a Droga , Glucurónidos/metabolismo , Glutatión/metabolismo , Humanos , Masculino , Ratas , Ratas Sprague-Dawley , Espectrometría de Masa por Ionización de Electrospray/métodos , Factores de Tiempo
13.
Drug Metab Dispos ; 31(11): 1327-36, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14570764

RESUMEN

Diclofenac, a nonsteroidal anti-inflammatory drug, is metabolized to a reactive acyl glucuronide that has been proposed to mediate toxic adverse drug reactions associated with its use. In the present study, we examined the ability of diclofenac acyl glucuronide (D-1-O-G) to transacylate glutathione (GSH) in vitro in buffer and in vivo in rats. Thus, in vitro reactions of D-1-O-G (100 microM) with GSH (10 mM) at pH 7.4 and 37 degrees C showed a linear time-dependent formation of diclofenac-S-acyl-glutathione (D-SG, 3 microM/h) through 60 min of incubation, reaching a maximum of 3.7 microM after 2 h of incubation. The major reaction that occurred was acyl migration of D-1-O-G (t1/2, 54 min) to less reactive isomers. The D-SG thioester product was shown to be unstable by degrading primarily to 1-(2,6-dichlorophenyl)indolin-2-one and by hydrolysis to diclofenac. After administration of diclofenac to rats (200 mg/kg), bile was collected and analyzed for D-SG by liquid chromatography-tandem mass spectrometry. Results indicated the presence of D-SG, which was confirmed by coelution with synthetic standard and by its tandem mass spectrum. When the reactivity of D-SG (100 microM) was compared with D-1-O-G (100 microM) in vitro in reactions with N-acetylcysteine (NAC, 10 mM), results showed the quantitative reaction of D-SG with NAC after 30 min of incubation, whereas only approximately 1% of D-1-O-G reacted to form diclofenac-S-acyl-NAC at the same time point. Results from these studies indicate that GSH reacts with D-1-O-G in vitro, and presumably in vivo, to form D-SG, and that the product D-SG thioester is chemically more reactive in transacylation-type reactions than the D-1-O-G metabolite.


Asunto(s)
Bilis/metabolismo , Diclofenaco/metabolismo , Glucurónidos/metabolismo , Glutatión/metabolismo , Animales , Cromatografía Líquida de Alta Presión/métodos , Diclofenaco/análisis , Diclofenaco/química , Glucurónidos/análisis , Glucurónidos/química , Glutatión/análisis , Glutatión/química , Masculino , Ratas , Ratas Sprague-Dawley
14.
Drug Metab Dispos ; 31(11): 1429-36, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14570776

RESUMEN

Zomepirac (ZP), a nonsteroidal anti-inflammatory drug that was withdrawn from use, is metabolized to zomepirac-1-O-acyl-glucuronide (ZP-1-O-G), a chemically reactive conjugate that has been implicated in the toxicity of the drug. In the present studies, we investigated the ability of ZP to become bioactivated to reactive metabolites that transacylate glutathione (GSH) forming ZP-S-acyl-glutathione thioester (ZP-SG) in vitro and in vivo in rat. When ZP (100 microM) was incubated with rat hepatocytes, ZP-SG was detected in incubation extracts by a sensitive selected reaction monitoring liquid chromatography/tandem mass spectrometry (LC/MS-MS) technique. The initial formation rate of ZP-SG was rapid and reached a maximum concentration of 0.24 +/- 0.03 nM after 4 min of incubation, then decreased, in a fairly linear fashion, to 0.07 +/- 0.03 nM after 60 min of incubation. The product ZP-SG (1 microM) was shown to be unstable by undergoing rapid hydrolysis (apparent half-life approximately 0.8 min) in incubations with rat hepatocytes. After administration of ZP to a male Sprague-Dawley rat (100 mg/kg i.p.), bile was collected and analyzed for ZP-SG by LC/MS-MS. Results indicated the presence of ZP-SG in bile (6.7 microg excreted after 6 h of collection), which was confirmed by coelution with synthetic standard and by its tandem mass spectrum. Together, these results demonstrate that ZP becomes metabolically activated in vitro in rat hepatocytes and in vivo in rat to reactive acylating derivative(s), such as ZP-1-O-G, that transacylate GSH forming ZP-SG. Finally, we propose that ZP-SG thioester could be used as a marker derivative for mechanistic studies on the bioactivation of the drug.


Asunto(s)
Bilis/química , Glutatión/análisis , Hepatocitos/química , Tolmetina/análogos & derivados , Tolmetina/análisis , Animales , Bilis/metabolismo , Glutatión/química , Glutatión/farmacocinética , Hepatocitos/metabolismo , Masculino , Ratas , Ratas Sprague-Dawley , Tolmetina/química , Tolmetina/farmacocinética
15.
Arch Biochem Biophys ; 414(2): 303-11, 2003 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-12781783

RESUMEN

In addition to glutathione (GSH) conjugating activity, glutathione S-transferases (GSTs) catalyze "reverse" reactions, such as the hydrolysis of GSH thiol esters. Reverse reactions are of interest as potential tumor-directed pro-drug activation strategies and as mechanisms for tissue redistribution of carboxylate-containing drugs. However, the mechanism and specificity of GST-mediated GSH thiol ester hydrolysis are uncharacterized. Here, the GSH thiol esters of ethacrynic acid (E-SG) and several nonsteroidal antiinflammatory agents have been tested as substrates with human GSTs. The catalytic hydrolysis of these thiol esters appears to be a general property of GSTs. The hydrolysis of the thiol ester of E-SG was studied further with GSTA1-1 and GSTP1-1, as a model pro-drug with several possible fates for the hydrolysis products: competitive inhibition, covalent enzyme adduction, and sequential metabolism. In contrast to hydrolysis rates, significant isoform-dependent differences in the subsequent fate of the products ethacrynic acid and GSH were observed. At low [E-SG], only the GSTP1-1 efficiently catalyzed sequential metabolism, via a dissociative mechanism.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Glutatión Transferasa/metabolismo , Profármacos/metabolismo , Antineoplásicos/farmacología , Unión Competitiva , Catálisis , Cromatografía Liquida , Activación Enzimática , Esterasas/metabolismo , Ésteres/metabolismo , Ácido Etacrínico/metabolismo , Glutatión/metabolismo , Gutatión-S-Transferasa pi , Glutatión Transferasa/química , Humanos , Hidrólisis , Isoenzimas/química , Cinética , Metiltransferasas , Modelos Químicos , Espectrometría de Masa por Ionización de Electrospray , Compuestos de Sulfhidrilo/metabolismo , Factores de Tiempo
16.
Drug Metab Dispos ; 31(6): 727-30, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12756204

RESUMEN

In this study, we investigated the possible involvement of acyl-CoA, reactive intermediary metabolites of 2-arylpropionic acids (profens), in protein adduct formation in rat liver homogenate and in human serum albumin (HSA) in buffer. (RS)-[1-14C]-2-Phenylpropionic acid (14C-2-PPA, 1 mM) was incubated with rat liver homogenate (1.5 mg/ml) in the presence of cofactors of acyl-CoA formation (Mg2+, ATP, and CoA). Aliquots of the incubation mixture were analyzed for covalent binding and acyl-CoA formation over a 3-h period. High-performance liquid chromatographic analysis of the products from such incubations showed the presence of 2-phenylpropionyl-S-acyl-CoA (2-PPA-CoA), which was confirmed by coelution with authentic 2-PPA-CoA, as well as by mass spectrometry. In the same incubations, 2-PPA was shown to bind covalently to hepatic proteins in a time- and ATP-dependent fashion. Inhibition of 2-PPA-CoA formation by acyl-CoA synthetase inhibitors, such as palmitic acid, lauric acid, octanoic acid, and ibuprofen, markedly decreased the extent of covalent binding of 2-PPA to hepatic proteins. Results from these in vitro studies strongly suggest that acyl-CoA thioester derivatives are chemically reactive and are able to bind covalently to tissue proteins in vitro, and, therefore, may contribute significantly to covalent adduct formation of profen drugs in vivo.


Asunto(s)
Acilcoenzima A/metabolismo , Fenilpropionatos/metabolismo , Albúmina Sérica/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Biotransformación , Cromatografía Líquida de Alta Presión , Humanos , Técnicas In Vitro , Hígado/metabolismo , Masculino , Espectrometría de Masas , Unión Proteica , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
17.
Toxicol Appl Pharmacol ; 187(2): 101-9, 2003 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-12649042

RESUMEN

2,4-Dichlorophenoxyacetic acid (2,4-D) is a widely used broadleaf herbicide that has been associated with acute liver toxicity in exposed humans or animals. Chemically reactive metabolites of 2,4-D are proposed as mediators of 2,4-D-induced hepatotoxicity. The aim of the present study was to investigate a novel reactive metabolite of 2,4-D, namely 2,4-dichlorophenoxyacetyl-S-acyl-CoA (2,4-D-CoA), and to determine its involvement in 2,4-D covalent adduct formation. Thus, incubations of synthetic 2,4-D-CoA (106 microM) with GSH (1 mM) in phosphate buffer (pH 7.4) showed 2,4-D-CoA to be able to transacylate the cysteine sulfhydryl of GSH, resulting in the formation of 2,4-D-S-acyl-glutathione (2,4-D-SG) thioester and reaching a concentration of 65 microM after 1 h of incubation. Under similar conditions, 2,4-D-CoA was shown to covalently bind to nucleophilic groups on human serum albumin (HSA, 30 mg/ml), resulting in time-dependent 2,4-D-HSA covalent adduct formation that reached a maximum of 440 pmol/mg HSA after 1 h of incubation. In addition to these studies, incubations of [1-(14)C]2,4-D (1 mM) with rat hepatocytes showed a time-dependent covalent binding of 2,4-D to hepatocyte protein. Inhibition of acyl-CoA formation by trimethylacetic acid (2 mM) decreased the amount of covalent binding to protein in rat hepatocytes by 50%. These results indicate that 2,4-D-CoA thioester is a reactive metabolite of 2,4-D that may contribute to 2,4-D-protein adduct formation in vivo and therefore the associated hepatotoxicity.


Asunto(s)
Ácido 2,4-Diclorofenoxiacético/metabolismo , Herbicidas/metabolismo , Fenoxiacetatos/metabolismo , Ácido 2,4-Diclorofenoxiacético/química , Ácido 2,4-Diclorofenoxiacético/toxicidad , Acilcoenzima A/antagonistas & inhibidores , Acilcoenzima A/metabolismo , Animales , Butiratos/farmacología , Cromatografía Líquida de Alta Presión , Glutatión/química , Glutatión/metabolismo , Hepatocitos/metabolismo , Herbicidas/química , Herbicidas/toxicidad , Humanos , Masculino , Fenoxiacetatos/química , Unión Proteica/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Albúmina Sérica/química , Albúmina Sérica/metabolismo
18.
Chem Res Toxicol ; 15(10): 1309-17, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12387630

RESUMEN

Chemically reactive species formed from the metabolism of carboxylic acid-containing compounds have been proposed as mediators of their toxic side-effects. Two alternative metabolic pathways known to be involved in the generation of reactive acylating metabolites of carboxylic acids are acyl glucuronidation and acyl-CoA formation. Here, we present studies with 2-phenylpropionic acid focused on evaluating the relative abilities of acyl glucuronides versus acyl-CoA derivatives to transacylate the nucleophilic cysteinyl-thiol of glutathione. Thus, synthetic 2-phenylpropionyl-S-acyl-CoA (2-PPA-SCoA) and biosynthetic 2-phenylpropionyl-1-O-acyl glucuronide (2-PPA-1-O-G) were incubated separately, and at varying concentrations (15.6-500 nM as well as at 0.1 mM), with GSH (1, 5, and 10 mM) in buffer (pH 7.4, 37 degrees C), and formation of the transacylation product, 2-phenylpropionyl-S-acyl-glutathione (2-PPA-SG), was quantified by reverse-phase HPLC and LC-MS. HPLC analysis of the products from both the reaction of 2-PPA-SCoA and 2-PPA-1-O-G with GSH showed the presence of 2-PPA-SG, which was confirmed by coelution with authentic 2-PPA-SG as well as by its LC/MS mass spectrum. The formation of 2-PPA-SG was time- and concentration-dependent with a formation rate constant of (1.9 +/- 0.2) x 10(-2) M(-1) x s(-1) from reactions of GSH with 2-PPA-SCoA, and (2.7 +/- 0.4) x 10(-4) M(-1) x s(-1) from reactions of GSH with 2-PPA-1-O-G. Therefore, the reactivity of 2-PPA-SCoA with GSH was 70 times greater than the reactivity of GSH with 2-PPA-1-O-G, which was found to acyl-migrate to less reactive isomers. Analysis of the in vitro stability of 2-PPA-SCoA and 2-PPA-1-O-G in the absence of GSH showed the CoA esters to be completely stable after 24 h, whereas the acyl glucuronides decomposed by 50% in 1.3 and 2.4 h of incubation at pH 7.4 and 37 degrees C for (R)- and (S)-2-PPA-1-O-G, respectively. In addition, studies of the reactivity of 2-PPA-SCoA with bovine serum albumin showed time- and pH-dependent covalent binding to the protein in vitro. These results support the hypothesis that xenobiotic acyl-CoA thioesters are reactive acylating species that, in addition to acyl glucuronides, may contribute to xenobiotic acid-protein adduct formation in vivo.


Asunto(s)
Acilcoenzima A/farmacología , Glucurónidos/farmacología , Fenilpropionatos/farmacología , Acilcoenzima A/química , Acilcoenzima A/metabolismo , Animales , Cromatografía Líquida de Alta Presión , Relación Dosis-Respuesta a Droga , Ésteres/química , Glucurónidos/química , Glucurónidos/metabolismo , Glutatión/metabolismo , Concentración de Iones de Hidrógeno , Masculino , Espectrometría de Masas , Fenilpropionatos/química , Fenilpropionatos/metabolismo , Proteínas/metabolismo , Ratas , Ratas Sprague-Dawley
19.
Drug Metab Dispos ; 30(1): 55-62, 2002 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11744612

RESUMEN

Clofibric acid (p-chlorophenoxyisobutyric acid) is metabolized in vivo to a thioester-linked glutathione conjugate, S-(p-chlorophenoxyisobutyryl)glutathione (CA-SG). The formation of this metabolite is presumed to occur via transacylation reactions between glutathione (GSH) and reactive acyl-linked metabolite(s) of the drug. The present study examines the chemical reactivity of clofibryl-S-acyl-CoA (CA-SCoA), an acyl-CoA thioester intermediary metabolite of clofibric acid, with GSH to form the CA-SG in vitro. Incubations of CA-SCoA (1 mM) with GSH (5 mM) were carried out at pH 7.5 and 37 degrees C, with analysis of the formed reaction products by isocratic reverse-phase high-performance liquid chromatography (HPLC). Results showed a time-dependent and linear formation of CA-SG up to 4 h (50 microM CA-SG formed/h), and after a 1-day incubation, the reaction mixture contained 0.7 mM CA-SG. The identity of CA-SG was confirmed by analysis of HPLC-purified material by tandem mass spectrometry. The rate of CA-SG formation was found to be increased 3-fold in incubations containing rat liver glutathione S-transferases (4 mg/ml). Analysis of the chemical stability of CA-SCoA in buffer at 37 degrees C and varying pH showed the derivative to be stable under mildly acidic and basic aqueous conditions but to hydrolyze at pH values greater than 10 after a 1-day incubation (t(1/2) = approximately 1 day at pH 10.5). Results from these studies show that CA-SCoA is a reactive thioester derivative of clofibric acid and is able to acylate GSH and other thiol-containing nucleophiles in vitro and, therefore, may be able to acylate protein thiols in vivo, which could contribute to the toxic side effects of the drug.


Asunto(s)
Acilcoenzima A/química , Ácido Clofíbrico/análogos & derivados , Ácido Clofíbrico/química , Glutatión/análogos & derivados , Glutatión/química , Xenobióticos/química , Acetilcisteína/química , Acilación , Animales , Cromatografía Líquida de Alta Presión , Ditiotreitol/química , Concentración de Iones de Hidrógeno , Ratas , Espectrometría de Masa por Ionización de Electrospray
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA